EP4159204B1 - Novel pharmaceutical composition - Google Patents

Novel pharmaceutical composition Download PDF

Info

Publication number
EP4159204B1
EP4159204B1 EP22209104.3A EP22209104A EP4159204B1 EP 4159204 B1 EP4159204 B1 EP 4159204B1 EP 22209104 A EP22209104 A EP 22209104A EP 4159204 B1 EP4159204 B1 EP 4159204B1
Authority
EP
European Patent Office
Prior art keywords
compound
tablet
suitably
tablets
tablet according
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP22209104.3A
Other languages
German (de)
English (en)
French (fr)
Other versions
EP4159204A1 (en
Inventor
Douglas J. DEMARIN
Ngocdiep T. Le
Francisco HENRIQUEZ
Lihong Wang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Novartis AG
Original Assignee
Novartis AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=46314827&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=EP4159204(B1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Novartis AG filed Critical Novartis AG
Publication of EP4159204A1 publication Critical patent/EP4159204A1/en
Application granted granted Critical
Publication of EP4159204B1 publication Critical patent/EP4159204B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2059Starch, including chemically or physically modified derivatives; Amylose; Amylopectin; Dextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61JCONTAINERS SPECIALLY ADAPTED FOR MEDICAL OR PHARMACEUTICAL PURPOSES; DEVICES OR METHODS SPECIALLY ADAPTED FOR BRINGING PHARMACEUTICAL PRODUCTS INTO PARTICULAR PHYSICAL OR ADMINISTERING FORMS; DEVICES FOR ADMINISTERING FOOD OR MEDICINES ORALLY; BABY COMFORTERS; DEVICES FOR RECEIVING SPITTLE
    • A61J3/00Devices or methods specially adapted for bringing pharmaceutical products into particular physical or administering forms
    • A61J3/06Devices or methods specially adapted for bringing pharmaceutical products into particular physical or administering forms into the form of pills, lozenges or dragees
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61JCONTAINERS SPECIALLY ADAPTED FOR MEDICAL OR PHARMACEUTICAL PURPOSES; DEVICES OR METHODS SPECIALLY ADAPTED FOR BRINGING PHARMACEUTICAL PRODUCTS INTO PARTICULAR PHYSICAL OR ADMINISTERING FORMS; DEVICES FOR ADMINISTERING FOOD OR MEDICINES ORALLY; BABY COMFORTERS; DEVICES FOR RECEIVING SPITTLE
    • A61J3/00Devices or methods specially adapted for bringing pharmaceutical products into particular physical or administering forms
    • A61J3/10Devices or methods specially adapted for bringing pharmaceutical products into particular physical or administering forms into the form of compressed tablets
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2009Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2095Tabletting processes; Dosage units made by direct compression of powders or specially processed granules, by eliminating solvents, by melt-extrusion, by injection molding, by 3D printing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2813Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present invention relates to tablets comprising N- ⁇ 3-[3-cyclopropyl-5-(2-fluoro-4-iodo-phenylamino)-6,8-dimethyl-2,4,7-trioxo-3,4,6,7-tetrahydro-2H-pyrido[4,3-d]pyrimidin-1-yl]phenyl ⁇ acetamide dimethyl sulfoxide solvate, represented by the following formula (I) and hereinafter referred to as Compound A:
  • Compound B is a compound which is disclosed and claimed, along with pharmaceutically acceptable salts and solvates thereof, as being useful as an inhibitor of MEK activity, particularly in treatment of cancer, in International Application No. PCT/JP2005/011082, having an International filing date of June 10, 2005 ; International Publication Number WO 2005/121142 and an International Publication date of December 22, 2005 .
  • Compound B is the compound of Example 4-1.
  • Compound B can be prepared as described in International Application No. PCT/JP2005/011082 .
  • Compound B can be prepared as described in United States Patent Publication No. US 2006/0014768, Published January 19, 2006 .
  • Compound B is the compound of Example 4-1.
  • Compound B is in the form of a dimethyl sulfoxide solvate, or Compound A as defined herein.
  • Compound B is in the form of a solvate selected from: hydrate, acetic acid, ethanol, nitromethane, chlorobenzene, 1-pentanol, isopropyl alcohol, ethylene glycol and 3-methyl-1-butanol.
  • Solvates and salt forms can be prepared by one of skill in the art, for example from the description in International Application No. PCT/JP2005/011082 or United States Patent Publication No. US 2006/0014768 .
  • Compound A is prepared in Example 4-149 of United States Patent Publication No. US 2006/0014768 .
  • Compound B is also disclosed as a MEK inhibitor in EP 2 125 810 A2 .
  • Solid oral pharmaceutical dosage forms are popular and useful forms of medications for dispensing pharmaceutically active compounds.
  • a variety of such forms are known, including tablets, capsules, pellets, lozenges, and powders.
  • each pharmaceutical compound acts uniquely in regards to therapeutic drug levels.
  • pharmaceutically active compounds particularly anti-neoplastic compounds, are often associated with undesirable side effects such as; toxicity (e.g. genotoxicity, teratogenicity) and undesirable physical or psychological manifestations.
  • the drug In addition to balancing the drug's unique chemical properties with those of the excipients, the drug must be administered at a specific amount that is sufficient to provide the desired therapeutic drug level but less than the amount that presents an unacceptable side effect profile, or within the therapeutic window for that particular drug.
  • the formulation and process of manufacture must be such as to provide an integral solid dosage form that maintains its integrity until used.
  • the solid dosage form must also possess acceptable dissolution and disintegration properties so as to provide the desired profile in use.
  • Pharmaceutically active compounds with low solubility and/or in solvate form can present particular challenges in preparing high quality solid dosage forms. These challenges include insufficient and in consistent exposure upon in vivo administration and desolvation which releases unsolvated compound which can exhibit poor pharmacodynamic properties.
  • the present invention relates to tablets comprising a therapeutically effective amount of Compound A, wherein the tablet is coated with a film coat containing a colored pigment, or wherein the tablet is an uncoated tablet, a wax coated tablet, or a white coated tablet placed in a protective light resistant blister pack or in a light resistant bottle.
  • the invention also relates to a process for making tablets comprising Compound A.
  • Another aspect of this invention relates to tablets comprising Compound A that are formulated using excipients, suitably the diluent component, that are substantially free of water, which as used herein and in the claims includes anhydrous versions of nonanhydrous excipients.
  • Such tablets exhibit improved properties. Such improved properties help to ensure safe and effective treatment.
  • Another aspect of this disclosure relates to a pharmaceutical tablet comprising a therapeutically effective amount of Compound A, wherein the tablet is prepared by compression of dry blend, suitably by direct compression or by dry granulation.
  • Such pharmaceutical tablet exhibits improved properties. Such improved properties help to ensure safe and effective treatment.
  • the invention also relates to a method of making direct compression and dry granulation pharmaceutical tablets comprising Compound A.
  • Another aspect of this invention relates to film coated oral pharmaceutical tablets comprising Compound A, wherein the film coat is an aqueous film coat composition comprising a film-forming polymer and water as a vehicle, suitably containing an iron oxide containing pigment or colorant.
  • Such tablets exhibit improved properties. Such improved properties help to ensure safe and effective treatment.
  • Another aspect of this disclosure relates to tablets comprising Compound A in an amount selected from: 0.5, 1 and 2mg, by weight of Compound B, wherein the tablet is coated with a film coat containing a colored pigment, or wherein the tablet is an uncoated tablet, a wax coated tablet, or a white coated tablet placed in a protective light resistant blister pack or in a light resistant bottle.
  • Such tablets exhibit improved properties. Such improved properties help to ensure safe and effective treatment.
  • Another aspect of this invention relates to tablets containing Compound A in which Compound A is in micronized form, wherein the tablet is coated with a film coat containing a colored pigment, or wherein the tablet is an uncoated tablet, a wax coated tablet, or a white coated tablet placed in a protective light resistant blister pack or in a light resistant bottle.
  • Such tablets exhibit improved properties. Such improved properties help to ensure safe and effective treatment.
  • Another aspect of this disclosure relates to tablets containing Compound A in which at least 50% of the Compound A particles have a particle size of 30 micron or less, suitably at least 50% of the Compound A particles have a particle size of 10 micron or less, suitably at least 50% of the Compound A particles have a particle size of 5 micron or less, wherein the tablet is coated with a film coat containing a colored pigment, or wherein the tablet is an uncoated tablet, a wax coated tablet, or a white coated tablet placed in a protective light resistant blister pack or in a light resistant bottle.
  • Such tablets exhibit improved properties. Such improved properties help to ensure safe and effective treatment.
  • Another aspect of this disclosure relates to tablets containing Compound A in which the amount of unsolvated compound (or Compound B as used herein) does not exceed about 20%, suitably the amount of unsolvated compound does not exceed about 15%, suitably the amount of unsolvated compound does not exceed about 10%, suitably the amount of unsolvated compound does not exceed about 5%, suitably the amount of unsolvated compound does not exceed about 2%, wherein the tablet is coated with a film coat containing a colored pigment, or wherein the tablet is an uncoated tablet, a wax coated tablet, or a white coated tablet placed in a protective light resistant blister pack or in a light resistant bottle.
  • Such tablets exhibit improved properties. Such improved properties help to ensure safe and effective treatment.
  • a solid oral pharmaceutical dosage form suitably a tablet, suitably a capsule, that contains an amount of Compound A selected from: 0.5, 1 and 2mg, by weight of Compound B.
  • a solid oral pharmaceutical dosage form suitably a tablet, suitably a capsule, that contains an amount of Compound A selected from: 0.5, 1 and 2mg, by weight of Compound B.
  • Compound A presents the formulator with unique concerns when attempting to formulate this compound into a suitable solid oral pharmaceutical dosage form, suitably a tablet within the therapeutic window for Compound A, particularly on a commercial scale.
  • Such concerns include but are not limited to; the tendency of the compound to revert to an insoluble desolvated form when exposed to moisture during the formulation process, slow dissolution of the compound from solid dosage forms, and that Compound A can suffer from photo-instability.
  • the present invention is directed to tablets that contain Compound A, suitably these tablets are produced on a commercial scale.
  • Compound A can suffer from photo-instability.
  • the potential for unacceptable levels of photo-degradation is of particular importance since photo-catalyzed degradation products may be potentially toxic.
  • Compound A tablets that are coated with an aqueous colored film coat suitably an iron oxide containing colored film coat, for example Opadry ® yellow or pink, exhibit improved photo-stability.
  • This improved stability leads to a reduction in the levels of photo-catalyzed degradation products forming upon light exposure. Such improved stability helps to ensure safe and effective treatment.
  • the present invention is directed to tablets containing Compound A that are coated with an aqueous colored film coat.
  • these tablet forms are produced on a commercial scale. These tablet forms help provide safe and effective treatment.
  • Compound A can cause toxic effects when administered in high doses. It has been discovered that Compound A, when administered in an amount selected from about 0.5mg, 1mg and 2mg, based on the amount of Compound B, is sufficient to provide the desired therapeutic drug level but less than the amount that presents an unacceptable side effect profile, or within the therapeutic window for Compound A.
  • the present invention is directed to tablets containing Compound A in an amount selected from: about 0.5mg, 1mg and 2mg based on the amount of Compound B. These tablet strengths help provide safe and effective treatment.
  • Compound A can undergo desolvation during handling and formulation resulting in unsolvated Compound B being formed.
  • Compound B is much less soluble than Compound A, which negatively impacts its pharmacodynamics when released from a pharmaceutical composition.
  • pharmaceutical formulations suitably tablets, suitably capsules, in which the amount of desolvated Compound B does not exceed 20%, suitably does not exceed 15%, suitably does not exceed 10%, suitably does not exceed 5%, suitably does not exceed 2%, when compared to Compound A, provide an acceptable release/pharmacodynamic profile.
  • the present invention is directed to tablets containing Compound B in an amount that does not exceed about 20%, suitably about 15%, suitably about 10%, suitably about 5%, suitably about 2% of the amount of Compound A.
  • Such tablets help provide safe and effective treatment.
  • Compound A can exhibit poor exposure and absorption upon in vivo administration. It has been found that pharmaceutical formulations, suitably tablets, suitably capsules, in which Compound A is micronized, suitably where at least 50% or the particles of Compound A are 30 micron or less, suitably at least 50% of the particles of Compound A are 10 micron or less, suitably at least 50% of the particles of Compound A are 5 micron or less, provide an acceptable exposure/absorption profile.
  • the present invention is directed to tablets containing Compound A in micronized form, suitably where at least 50% of the particles of Compound A are 30 micron or less, suitably at least 50% of the particles of Compound A are 10 micron or less, suitably at least 50% of the particles of Compound A are 5 micron or less.
  • Such tablets help provide safe and effective treatment.
  • improved properties contemplates several advantages to the pharmacokinetic profile of the in vivo release of Compound A from a formulation, suitably a solid oral pharmaceutical dosage form, a tablet, that utilizes an aspect of the present invention when compared to a formulation that does not utilize that aspect of the present invention, suitably the formulation is produced on a commercial scale.
  • improved properties include: increased oral bioavailability, improved physical and chemical stability, improved photo-stability, a consistent pharmacokinetic profile, an improved pharmacokinetic profile and a consistent dissolution rate.
  • drug or “active ingredient” and derivatives thereof, unless otherwise defined, means Compound A or N- ⁇ 3-[3-cyclopropyl-5-(2-fluoro-4-iodo-phenylamino)-6,8-dimethyl-2,4,7-trioxo-3,4,6,7-tetrahydro-2H-pyrido[4,3-d]pyrimidin-1-yl]phenyl ⁇ acetamide dimethyl sulfoxide.
  • Compound B and derivatives thereof, means N- ⁇ 3-[3-cyclopropyl-5-(2-fluoro-4-iodo-phenylamino)-6,8-dimethyl-2,4,7-trioxo-3,4,6,7-tetrahydro-2H-pyrido[4,3-d]pyrimidin-1-yl]phenyl ⁇ acetamide, as the free or unsalted and unsolvated compound.
  • Compound B also refers to the amount of free or unsalted and unsolvated compound in an amount of Compound A.
  • commercial scale preparation of a batch scale greater than about 20 kg of direct compression mix, suitably greater than 50 kg, suitably greater than 75 kg or a batch size suitable to prepare at least about 50,000 solid oral pharmaceutical dosage forms, suitably tablets, suitably capsules, suitably at least 75,000 solid oral pharmaceutical dosage forms, suitably tablets, suitably capsules, suitably at least 100,000 solid oral pharmaceutical dosage forms, suitably tablets, suitably capsules.
  • an effective amount means that amount of a drug or active ingredient that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for instance, by a researcher or clinician.
  • therapeutically effective amount means any amount which, as compared to a corresponding subject who has not received such amount, results in improved treatment, healing, prevention, or amelioration of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder.
  • the term also includes within its scope amounts effective to enhance normal physiological function.
  • formulation refers to solid oral pharmaceutical dosage forms, suitably tablets, suitably capsules, of the invention that contain Compound A.
  • co-administration is meant either simultaneous administration or any manner of separate sequential administration of a solid oral pharmaceutical dosage form containing Compound A, and a further active agent or agents, known to be useful in the treatment of cancer, including chemotherapy and radiation treatment.
  • further active agent or agents includes any compound or therapeutic agent known to or that demonstrates advantageous properties when administered to a patient in need of treatment for cancer.
  • further active agent or agents is used interchangeably with further anti-neoplastic agent or agents.
  • the compounds are administered in a close time proximity to each other.
  • one compound may be administered by injection and another compound may be administered orally.
  • the "co-administration” will consist essentially of a solid oral pharmaceutical dosage form containing compound A and a second pharmaceutical dosage form containing a further active agent.
  • the "co-administration” will consist essentially of a solid oral pharmaceutical dosage form containing compound A, a second pharmaceutical dosage form containing a further active agent, and a third pharmaceutical dosage form containing another further active agent.
  • any anti-neoplastic agent that has activity versus a susceptible tumor being treated may be co-administered in the treatment of cancer in the present invention.
  • examples of such agents can be found in Cancer Principles and Practice of Oncology by V.T. Devita and S. Hellman (editors), 6th edition (February 15, 2001), Lippincott Williams & Wilkins Publishers .
  • a person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the cancer involved.
  • Typical anti-neoplastic agents useful in the present invention include, but are not limited to, anti-microtubule agents such as diterpenoids and vinca alkaloids; platinum coordination complexes; alkylating agents such as nitrogen mustards, oxazaphosphorines, alkylsulfonates, nitrosoureas, and triazenes; antibiotic agents such as anthracyclins, actinomycins and bleomycins; topoisomerase II inhibitors such as epipodophyllotoxins; antimetabolites such as purine and pyrimidine analogues and anti-folate compounds; topoisomerase I inhibitors such as camptothecins; hormones and hormonal analogues; signal transduction pathway inhibitors; non-receptor tyrosine kinase angiogenesis inhibitors; immunotherapeutic agents; proapoptotic agents; cell cycle signaling inhibitors; proteasome inhibitors; and inhibitors of cancer metabolism.
  • anti-microtubule agents such as
  • Examples of a further active agent or agents (anti-neoplastic agent) for use in combination or co-administered with a presently invented tablet, are chemotherapeutic agents.
  • Anti-microtubule or anti-mitotic agents are phase specific agents active against the microtubules of tumor cells during M or the mitosis phase of the cell cycle.
  • anti-microtubule agents include, but are not limited to, diterpenoids and vinca alkaloids.
  • Diterpenoids which are derived from natural sources, are phase specific anticancer agents that operate at the G 2 /M phases of the cell cycle. It is believed that the diterpenoids stabilize the ⁇ -tubulin subunit of the microtubules, by binding with this protein. Disassembly of the protein appears then to be inhibited with mitosis being arrested and cell death following. Examples of diterpenoids include, but are not limited to, paclitaxel and its analog docetaxel.
  • Paclitaxel 5 ⁇ ,20-epoxy-1,2 ⁇ ,4,7 ⁇ ,10 ⁇ , 13 ⁇ -hexa-hydroxytax-11-en-9-one 4,10-diacetate 2-benzoate 13-ester with (2R,3S)-N-benzoyl-3-phenylisoserine; is a natural diterpene product isolated from the Pacific yew tree Taxus brevifolia and is commercially available as an injectable solution TAXOL ® . It is a member of the taxane family of terpenes. It was first isolated in 1971 by Wani et al. J. Am. Chem, Soc., 93:2325. 1971 ), who characterized its structure by chemical and X-ray crystallographic methods.
  • Paclitaxel has been approved for clinical use in the treatment of refractory ovarian cancer in the United States ( Markman et al., Yale Journal of Biology and Medicine, 64:583, 1991 ; McGuire et al., Ann. intern, Med., 111:273,1989 ) and for the treatment of breast cancer ( Holmes et al., J. Nat. Cancer Inst., 83:1797,1991 .) It is a potential candidate for treatment of neoplasms in the skin ( Einzig et. al., Proc. Am. Soc. Clin. Oncol., 20:46 ) and head and neck carcinomas ( Forastire et. al., Sem. Oncol., 20:56, 1990 ).
  • the compound also shows potential for the treatment of polycystic kidney disease ( Woo et. al., Nature, 368:750. 1994 ), lung cancer and malaria.
  • Treatment of patients with paclitaxel results in bone marrow suppression (multiple cell lineages, Ignoff, R.J. et. al, Cancer Chemotherapy Pocket Guide 1998 ) related to the duration of dosing above a threshold concentration (50nM) ( Kearns, C.M. et. al., Seminars in Oncology, 3(6) p.16-23, 1995 ).
  • Docetaxel (2R,3S)- N-carboxy-3-phenylisoserine,N-tert-butyl ester, 13-ester with 5 ⁇ -20-epoxy-1,2 ⁇ ,4,7 ⁇ ,10 ⁇ ,13 ⁇ -hexahydroxytax-11-en-9-one 4-acetate 2-benzoate, trihydrate; is commercially available as an injectable solution as TAXOTERE ® .
  • Docetaxel is indicated for the treatment of breast cancer.
  • Docetaxel is a semisynthetic derivative of paclitaxel q.v., prepared using a natural precursor, 10-deacetyl-baccatin III, extracted from the needle of the European Yew tree. The dose limiting toxicity of docetaxel is neutropenia.
  • Vinca alkaloids are phase specific anti-neoplastic agents derived from the periwinkle plant. Vinca alkaloids act at the M phase (mitosis) of the cell cycle by binding specifically to tubulin. Consequently, the bound tubulin molecule is unable to polymerize into microtubules. Mitosis is believed to be arrested in metaphase with cell death following. Examples of vinca alkaloids include, but are not limited to, vinblastine, vincristine, and vinorelbine.
  • Vinblastine vincaleukoblastine sulfate
  • VELBAN ® an injectable solution
  • Myelosuppression is the dose limiting side effect of vinblastine.
  • Vincristine vincaleukoblastine, 22-oxo-, sulfate
  • ONCOVIN ® an injectable solution.
  • Vincristine is indicated for the treatment of acute leukemias and has also found use in treatment regimens for Hodgkin's and non-Hodgkin's malignant lymphomas.
  • Alopecia and neurologic effects are the most common side effect of vincristine and to a lesser extent myelosupression and gastrointestinal mucositis effects occur.
  • Vinorelbine 3',4'-didehydro -4'-deoxy-C'-norvincaleukoblastine [R-(R*,R*)-2,3-dihydroxybutanedioate (1:2)(salt)], commercially available as an injectable solution of vinorelbine tartrate (NAVELBINE ® ), is a semisynthetic vinca alkaloid.
  • Vinorelbine is indicated as a single agent or in combination with other chemotherapeutic agents, such as cisplatin, in the treatment of various solid tumors, particularly non-small cell lung, advanced breast, and hormone refractory prostate cancers. Myelosuppression is the most common dose limiting side effect of vinorelbine.
  • Platinum coordination complexes are non-phase specific anti-cancer agents, which are interactive with DNA.
  • the platinum complexes enter tumor cells, undergo, aquation and form intra- and interstrand crosslinks with DNA causing adverse biological effects to the tumor.
  • Examples of platinum coordination complexes include, but are not limited to, cisplatin and carboplatin.
  • Cisplatin cis-diamminedichloroplatinum
  • PLATINOL ® an injectable solution.
  • Cisplatin is primarily indicated in the treatment of metastatic testicular and ovarian cancer and advanced bladder cancer.
  • the primary dose limiting side effects of cisplatin are nephrotoxicity, which may be controlled by hydration and diuresis, and ototoxicity.
  • Carboplatin platinum, diammine [1,1-cyclobutane-dicarboxylate(2-)-O,O'], is commercially available as PARAPLATIN ® as an injectable solution.
  • Carboplatin is primarily indicated in the first and second line treatment of advanced ovarian carcinoma. Bone marrow suppression is the dose limiting toxicity of carboplatin.
  • Alkylating agents are non-phase anti-cancer specific agents and strong electrophiles. Typically, alkylating agents form covalent linkages, by alkylation, to DNA through nucleophilic moieties of the DNA molecule such as phosphate, amino, sulfhydryl, hydroxyl, carboxyl, and imidazole groups. Such alkylation disrupts nucleic acid function leading to cell death.
  • alkylating agents include, but are not limited to, nitrogen mustards such as cyclophosphamide, melphalan, and chlorambucil; alkyl sulfonates such as busulfan; nitrosoureas such as carmustine; and triazenes such as dacarbazine.
  • Cyclophosphamide 2-[bis(2-chloroethyl)amino]tetrahydro-2H-1,3,2-oxazaphosphorine 2-oxide monohydrate, is commercially available as an injectable solution or tablets as CYTOXAN ® . Cyclophosphamide is indicated as a single agent or in combination with other chemotherapeutic agents, in the treatment of malignant lymphomas, multiple myeloma, and leukemias. Alopecia, nausea, vomiting and leukopenia are the most common dose limiting side effects of cyclophosphamide.
  • Melphalan 4-[bis(2-chloroethyl)amino]-L-phenylalanine, is commercially available as an injectable solution or tablets as ALKERAN ® .
  • Melphalan is indicated for the palliative treatment of multiple myeloma and non-resectable epithelial carcinoma of the ovary. Bone marrow suppression is the most common dose limiting side effect of melphalan.
  • Chlorambucil 4-[bis(2-chloroethyl)amino]benzenebutanoic acid, is commercially available as LEUKERAN ® tablets. Chlorambucil is indicated for the palliative treatment of chronic lymphatic leukemia, and malignant lymphomas such as lymphosarcoma, giant follicular lymphoma, and Hodgkin's disease. Bone marrow suppression is the most common dose limiting side effect of chlorambucil.
  • Busulfan 1,4-butanediol dimethanesulfonate, is commercially available as MYLERAN ® TABLETS. Busulfan is indicated for the palliative treatment of chronic myelogenous leukemia. Bone marrow suppression is the most common dose limiting side effects of busulfan.
  • Carmustine 1,3-[bis(2-chloroethyl)-1-nitrosourea, is commercially available as single vials of lyophilized material as BiCNU ® .
  • Carmustine is indicated for the palliative treatment as a single agent or in combination with other agents for brain tumors, multiple myeloma, Hodgkin's disease, and non-Hodgkin's lymphomas. Delayed myelosuppression is the most common dose limiting side effects of carmustine.
  • dacarbazine 5-(3,3-dimethyl-1-triazeno)-imidazole-4-carboxamide, is commercially available as single vials of material as DTIC-Dome ® .
  • dacarbazine is indicated for the treatment of metastatic malignant melanoma and in combination with other agents for the second line treatment of Hodgkin's Disease. Nausea, vomiting, and anorexia are the most common dose limiting side effects of dacarbazine.
  • Antibiotic anti-neoplastics are non-phase specific agents, which bind or intercalate with DNA. Typically, such action results in stable DNA complexes or strand breakage, which disrupts ordinary function of the nucleic acids leading to cell death.
  • antibiotic anti-neoplastic agents include, but are not limited to, actinomycins such as dactinomycin, anthrocyclins such as daunorubicin and doxorubicin; and bleomycins.
  • Dactinomycin also know as Actinomycin D, is commercially available in injectable form as COSMEGEN ® . Dactinomycin is indicated for the treatment of Wilm's tumor and rhabdomyosarcoma. Nausea, vomiting, and anorexia are the most common dose limiting side effects of dactinomycin.
  • Daunorubicin (8S-cis-)-8-acetyl-10-[(3-amino-2,3,6-trideoxy- ⁇ -L-lyxo-hexopyranosyl)oxy]-7,8,9,10-tetrahydro-6,8,11-trihydroxy-1-methoxy-5,12 naphthacenedione hydrochloride, is commercially available as a liposomal injectable form as DAUNOXOME ® or as an injectable as CERUBIDINE ® . Daunorubicin is indicated for remission induction in the treatment of acute nonlymphocytic leukemia and advanced HIV associated Kaposi's sarcoma. Myelosuppression is the most common dose limiting side effect of daunorubicin.
  • Doxorubicin (8S, 10S)-10-[(3-amino-2,3,6-trideoxy- ⁇ -L-lyxo-hexopyranosyl)oxy]-8-glycoloyl, 7,8,9,10-tetrahydro-6,8,11-trihydroxy-1-methoxy-5,12 naphthacenedione hydrochloride, is commercially available as an injectable form as RUBEX ® or ADRIAMYCIN RDF ® .
  • Doxorubicin is primarily indicated for the treatment of acute lymphoblastic leukemia and acute myeloblastic leukemia, but is also a useful component in the treatment of some solid tumors and lymphomas. Myelosuppression is the most common dose limiting side effect of doxorubicin.
  • Bleomycin a mixture of cytotoxic glycopeptide antibiotics isolated from a strain of Streptomyces verticillus, is commercially available as BLENOXANE ® .
  • Bleomycin is indicated as a palliative treatment, as a single agent or in combination with other agents, of squamous cell carcinoma, lymphomas, and testicular carcinomas. Pulmonary and cutaneous toxicities are the most common dose limiting side effects of bleomycin.
  • Topoisomerase II inhibitors include, but are not limited to, epipodophyllotoxins.
  • Epipodophyllotoxins are phase specific anti-neoplastic agents derived from the mandrake plant. Epipodophyllotoxins typically affect cells in the S and G 2 phases of the cell cycle by forming a ternary complex with topoisomerase II and DNA causing DNA strand breaks. The strand breaks accumulate and cell death follows. Examples of epipodophyllotoxins include, but are not limited to, etoposide and teniposide.
  • Etoposide 4'-demethyl-epipodophyllotoxin 9[4,6-0-(R)-ethylidene- ⁇ -D-glucopyranoside]
  • VePESID ® an injectable solution or capsules
  • VP-16 an injectable solution or capsules
  • Etoposide is indicated as a single agent or in combination with other chemotherapy agents in the treatment of testicular and non-small cell lung cancers. Myelosuppression is the most common side effect of etoposide. The incidence of leucopenia tends to be more severe than thrombocytopenia.
  • Teniposide 4'-demethyl-epipodophyllotoxin 9[4,6-0-(R)-thenylidene- ⁇ -D-glucopyranoside], is commercially available as an injectable solution as VUMON ® and is commonly known as VM-26.
  • Teniposide is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia in children. Myelosuppression is the most common dose limiting side effect of teniposide.
  • Teniposide can induce both leucopenia and thrombocytopenia.
  • Antimetabolite neoplastic agents are phase specific anti-neoplastic agents that act at S phase (DNA synthesis) of the cell cycle by inhibiting DNA synthesis or by inhibiting purine or pyrimidine base synthesis and thereby limiting DNA synthesis. Consequently, S phase does not proceed and cell death follows.
  • Examples of antimetabolite anti-neoplastic agents include, but are not limited to, fluorouracil, methotrexate, cytarabine, mecaptopurine, thioguanine, and gemcitabine.
  • 5-fluorouracil 5-fluoro-2,4- (1H,3H) pyrimidinedione
  • fluorouracil is commercially available as fluorouracil.
  • Administration of 5-fluorouracil leads to inhibition of thymidylate synthesis and is also incorporated into both RNA and DNA. The result typically is cell death.
  • 5-fluorouracil is indicated as a single agent or in combination with other chemotherapy agents in the treatment of carcinomas of the breast, colon, rectum, stomach and pancreas. Myelosuppression and mucositis are dose limiting side effects of 5-fluorouracil.
  • Other fluoropyrimidine analogs include 5-fluoro deoxyuridine (floxuridine) and 5-fluorodeoxyuridine monophosphate.
  • Cytarabine 4-amino-1- ⁇ -D-arabinofuranosyl-2 (1H)-pyrimidinone, is commercially available as CYTOSAR-U ® and is commonly known as Ara-C. It is believed that cytarabine exhibits cell phase specificity at S-phase by inhibiting DNA chain elongation by terminal incorporation of cytarabine into the growing DNA chain. Cytarabine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Other cytidine analogs include 5-azacytidine and 2',2'-difluorodeoxycytidine (gemcitabine). Cytarabine induces leucopenia, thrombocytopenia, and mucositis.
  • Mercaptopurine 1,7-dihydro-6H-purine-6-thione monohydrate, is commercially available as PURINETHOL ® .
  • Mercaptopurine exhibits cell phase specificity at S-phase by inhibiting DNA synthesis by an as of yet unspecified mechanism.
  • Mercaptopurine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Myelosuppression and gastrointestinal mucositis are expected side effects of mercaptopurine at high doses.
  • a useful mercaptopurine analog is azathioprine.
  • Thioguanine 2-amino-1,7-dihydro-6H-purine-6-thione, is commercially available as TABLOID ® .
  • Thioguanine exhibits cell phase specificity at S-phase by inhibiting DNA synthesis by an as of yet unspecified mechanism.
  • Thioguanine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia.
  • Myelosuppression including leucopenia, thrombocytopenia, and anemia, is the most common dose limiting side effect of thioguanine administration. However, gastrointestinal side effects occur and can be dose limiting.
  • Other purine analogs include pentostatin, erythrohydroxynonyladenine, fludarabine phosphate, and cladribine.
  • Gemcitabine 2'-deoxy-2', 2'-difluorocytidine monohydrochloride ( ⁇ -isomer), is commercially available as GEMZAR ® .
  • Gemcitabine exhibits cell phase specificity at S-phase and by blocking progression of cells through the G1/S boundary.
  • Gemcitabine is indicated in combination with cisplatin in the treatment of locally advanced non-small cell lung cancer and alone in the treatment of locally advanced pancreatic cancer.
  • Myelosuppression including leucopenia, thrombocytopenia, and anemia, is the most common dose limiting side effect of gemcitabine administration.
  • Methotrexate N-[4[[(2,4-diamino-6-pteridinyl) methyl]methylamino] benzoyl]-L-glutamic acid, is commercially available as methotrexate sodium. Methotrexate exhibits cell phase effects specifically at S-phase by inhibiting DNA synthesis, repair and/or replication through the inhibition of dyhydrofolic acid reductase which is required for synthesis of purine nucleotides and thymidylate.
  • Methotrexate is indicated as a single agent or in combination with other chemotherapy agents in the treatment of choriocarcinoma, meningeal leukemia, non-Hodgkin's lymphoma, and carcinomas of the breast, head, neck, ovary and bladder.
  • Myelosuppression (leucopenia, thrombocytopenia, and anemia) and mucositis are expected side effect of methotrexate administration.
  • Camptothecins including, camptothecin and camptothecin derivatives are available or under development as Topoisomerase I inhibitors. Camptothecins cytotoxic activity is believed to be related to its Topoisomerase I inhibitory activity. Examples of camptothecins include, but are not limited to irinotecan, topotecan, and the various optical forms of 7-(4-methylpiperazino-methylene)-10,1 1-ethylenedioxy-20-camptothecin described below.
  • Irinotecan HCl (4S)-4,11-diethyl-4-hydroxy-9-[(4-piperidinopiperidino) carbonyloxy]-1H-pyrano[3',4',6,7]indolizino[1,2-b]quinoline-3,14(4H,12H)-dione hydrochloride, is commercially available as the injectable solution CAMPTOSAR ® .
  • Irinotecan is a derivative of camptothecin which binds, along with its active metabolite SN-38, to the topoisomerase I - DNA complex. It is believed that cytotoxicity occurs as a result of irreparable double strand breaks caused by interaction of the topoisomerase I : DNA : irintecan or SN-38 ternary complex with replication enzymes. Irinotecan is indicated for treatment of metastatic cancer of the colon or rectum. The dose limiting side effects of irinotecan HCI are myelosuppression, including neutropenia, and GI effects, including diarrhea.
  • Topotecan HCl (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1H-pyrano[3',4',6,7]indolizino[1,2-b]quinoline-3,14-(4H,12H)-dione monohydrochloride, is commercially available as the injectable solution HYCAMTIN ® .
  • Topotecan is a derivative of camptothecin which binds to the topoisomerase I - DNA complex and prevents religation of singles strand breaks caused by Topoisomerase I in response to torsional strain of the DNA molecule.
  • Topotecan is indicated for second line treatment of metastatic carcinoma of the ovary and small cell lung cancer.
  • the dose limiting side effect of topotecan HCI is myelosuppression, primarily neutropenia.
  • camptothecin derivative of Formula A including the racemic mixture (R,S) form as well as the R and S enantiomers: known by the chemical name "7-(4-methylpiperazino-methylene)-10,11-ethylenedioxy-20(R,S)-camptothecin (racemic mixture) or "7-(4-methylpiperazino-methylene)-10,11-ethylenedioxy-20(R)-camptothecin (R enantiomer) or "7-(4-methylpiperazino-methylene)-10,11-ethylenedioxy-20(S)-camptothecin (S enantiomer).
  • Such compound as well as related compounds are described, including methods of making, in U.S. Patent Nos. 6,063,923 ; 5,342,947 ; 5,559,235 ; 5,491,237 and pending U.S. patent Application No. 08/977,217 filed November 24, 1997 .
  • Hormones and hormonal analogues are useful compounds for treating cancers in which there is a relationship between the hormone(s) and growth and/or lack of growth of the cancer.
  • hormones and hormonal analogues useful in cancer treatment include, but are not limited to, adrenocorticosteroids such as prednisone and prednisolone which are useful in the treatment of malignant lymphoma and acute leukemia in children; aminoglutethimide and other aromatase inhibitors such as anastrozole, letrazole, vorazole, and exemestane useful in the treatment of adrenocortical carcinoma and hormone dependent breast carcinoma containing estrogen receptors; progestrins such as megestrol acetate useful in the treatment of hormone dependent breast cancer and endometrial carcinoma; estrogens, androgens, and anti-androgens such as flutamide, nilutamide, bicalutamide, cyproterone acetate and 5 ⁇ -reductases
  • Patent Nos. 5,681,835 , 5,877,219 , and 6,207,716 useful in the treatment of hormone dependent breast carcinoma and other susceptible cancers; and gonadotropin-releasing hormone (GnRH) and analogues thereof which stimulate the release of leutinizing hormone (LH) and/or follicle stimulating hormone (FSH) for the treatment prostatic carcinoma, for instance, LHRH agonists and antagagonists such as goserelin acetate and luprolide.
  • GnRH gonadotropin-releasing hormone
  • LH leutinizing hormone
  • FSH follicle stimulating hormone
  • Signal transduction pathway inhibitors are those inhibitors, which block or inhibit a chemical process which evokes an intracellular change. As used herein this change is cell proliferation or differentiation.
  • Signal tranduction inhibitors useful in the present invention include inhibitors of receptor tyrosine kinases, non-receptor tyrosine kinases, SH2/SH3 domain blockers, serine/threonine kinases, phosphotidylinositol-3 kinases, myo-inositol signaling, and Ras oncogenes.
  • protein tyrosine kinases catalyse the phosphorylation of specific tyrosyl residues in various proteins involved in the regulation of cell growth.
  • protein tyrosine kinases can be broadly classified as receptor or non-receptor kinases.
  • Receptor tyrosine kinases are transmembrane proteins having an extracellular ligand binding domain, a transmembrane domain, and a tyrosine kinase domain. Receptor tyrosine kinases are involved in the regulation of cell growth and are generally termed growth factor receptors. Inappropriate or uncontrolled activation of many of these kinases, i.e. aberrant kinase growth factor receptor activity, for example by over-expression or mutation, has been shown to result in uncontrolled cell growth. Accordingly, the aberrant activity of such kinases has been linked to malignant tissue growth. Consequently, inhibitors of such kinases could provide cancer treatment methods.
  • Growth factor receptors include, for example, epidermal growth factor receptor (EGFr), platelet derived growth factor receptor (PDGFr), erbB2, erbB4, vascular endothelial growth factor receptor (VEGFr), tyrosine kinase with immunoglobulin-like and epidermal growth factor homology domains (TIE-2), insulin growth factor-I (IGFI) receptor, macrophage colony stimulating factor (cfms), BTK, ckit, cmet, fibroblast growth factor (FGF) receptors, Trk receptors (TrkA, TrkB, and TrkC), ephrin (eph) receptors, and the RET protooncogene.
  • EGFr epidermal growth factor receptor
  • PDGFr platelet derived growth factor receptor
  • erbB2 erbB4
  • VEGFr vascular endothelial growth factor receptor
  • TIE-2 vascular endothelial growth factor receptor
  • IGFI insulin growth factor
  • inhibitors of growth receptors include ligand antagonists, antibodies, tyrosine kinase inhibitors and anti-sense oligonucleotides.
  • Growth factor receptors and agents that inhibit growth factor receptor function are described, for instance, in Kath, John C., Exp. Opin. Ther. Patents (2000) 10(6):803-818 ; Shawver et al DDT Vol 2, No. 2 February 1997 ; and Lofts, F. J. et al, "Growth factor receptors as targets", New Molecular Targets for Cancer Chemotherapy, ed. Workman, Paul and Kerr, David, CRC press 1994, Lond on.
  • Non-receptor tyrosine kinases which are not growth factor receptor kinases are termed non-receptor tyrosine kinases.
  • Non-receptor tyrosine kinases for use in the present invention include cSrc, Lck, Fyn, Yes, Jak, cAbl, FAK (Focal adhesion kinase), Brutons tyrosine kinase, and Bcr-Abl.
  • Such non-receptor kinases and agents which inhibit non-receptor tyrosine kinase function are described in Sinh, S.
  • SH2/SH3 domain blockers are agents that disrupt SH2 or SH3 domain binding in a variety of enzymes or adaptor proteins including, PI3-K p85 subunit, Src family kinases, adaptor molecules (Shc, Crk, Nck, Grb2) and Ras-GAP.
  • SH2/SH3 domains as targets for anti-cancer drugs are discussed in Smithgall, T.E. (1995), Journal of Pharmacological and Toxicological Methods. 34(3) 125-32 .
  • Inhibitors of Serine/Threonine Kinases including MAP kinase cascade blockers which include blockers of Raf kinases (rafk), Mitogen or Extracellular Regulated Kinase (MEKs), and Extracellular Regulated Kinases (ERKs); and Protein kinase C family member blockers including blockers of PKCs (alpha, beta, gamma, epsilon, mu, lambda, iota, zeta).
  • IkB kinase family IKKa, IKKb
  • PKB family kinases akt kinase family members
  • PDK1 and TGF beta receptor kinases IkB kinase family
  • Serine/Threonine kinases and inhibitors thereof are described in Yamamoto, T., Taya, S., Kaibuchi, K., (1999), Journal of Biochemistry. 126 (5) 799-803 ; Brodt, P, Samani, A., and Navab, R. (2000), Biochemical Pharmacology, 60. 1101-1107 ; Massague, J., Weis-Garcia, F. (1996) Cancer Surveys. 27:41-64 ; Philip, P.A., and Harris, A.L. (1995), Cancer Treatment and Research. 78: 3-27 , Lackey, K. et al Bioorganic and Medicinal Chemistry Letters, (10), 2000, 223-226 ; U.S. Patent No. 6,268,391 ; Pearce, L.R et al. Nature Reviews Molecular Cell Biology (2010) 11, 9-22 . and Martinez-lacaci, L., et al, Int. J. Cancer (2000), 88(1), 44-52 .
  • the pharmaceutically active compound of the invention is used in combination with a B-Raf inhibitor.
  • a B-Raf inhibitor e.g., N- ⁇ 3-[5-(2-Amino-4-pyrimidinyl)-2-(1,1-dimethylethyl)-1,3-thiazol-4-yl]-2-fluorophenyl ⁇ -2,6-difluorobenzenesulfonamide, or a pharmaceutically acceptable salt thereof, which is disclosed and claimed, in International Application No. PCT/US2009/042682, having an International filing date of May 4, 2009 .
  • N - ⁇ 3-[5-(2-Amino-4-pyrimidinyl)-2-(1,1-dimethylethyl)-1,3-thiazol-4-yl]-2-fluorophenyl ⁇ -2,6-difluorobenzenesulfonamide can be prepared as described in International Application No. PCT/US2009/042682 .
  • the pharmaceutically active compound of the invention is used in combination with an Akt inhibitor.
  • an Akt inhibitor e.g., N- ⁇ (1S)-2-amino-1-[(3,4-difluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-furancarboxamide or a pharmaceutically acceptable salt thereof, which is disclosed and claimed in International Application No. PCT/US2008/053269, having an International filing date of February 7, 2008 ; International Publication Number WO 2008/098104 and an International Publication date of August 14, 2008 .
  • N- ⁇ (1S)-2-amino-1-[(3,4-difluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1 H-pyrazol-5-yl)-2-furancarboxamide is the compound of example 224 and can be prepared as described in International Application No. PCT/US2008/053269 .
  • the pharmaceutically active compound of the invention is used in combination with an Akt inhibitor.
  • an Akt inhibitor e.g., N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide or a pharmaceutically acceptable salt thereof, which is disclosed and claimed in International Application No. PCT/US2008/053269, having an International filing date of February 7, 2008 ; International Publication Number WO 2008/098104 and an International Publication date of August 14, 2008 .
  • N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide is the compound of example 96 and can be prepared as described in International Application No. PCT/US2008/053269 .
  • N- ⁇ (1S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1H-pyrazol-5-yl)-2-thiophenecarboxamide is in the form of a hydrochloride salt.
  • the salt form can be prepared by one of skill in the art from the description in International Application No. PCT/US2010/022323, having an International filing date of January 28, 2010 .
  • Inhibitors of Phosphotidylinositol-3 Kinase family members including blockers of PI3-kinase, ATM, DNA-PK, and Ku may also be useful in the present invention.
  • Such kinases are discussed in Abraham, R.T. (1996), Current Opinion in Immunology. 8 (3) 412-8 ; Canman, C.E., Lim, D.S. (1998), Oncogene 17 (25) 3301-3308 ; Jackson, S.P. (1997), International Journal of Biochemistry and Cell Biology. 29 (7):935-8 ; and Zhong, H. et al, Cancer res, (2000) 60(6), 1541-1545 .
  • Myo-inositol signaling inhibitors such as phospholipase C blockers and Myoinositol analogues.
  • signal inhibitors are described in Powis, G., and Kozikowski A., (1994) New Molecular Targets for Cancer Chemotherapy ed., Paul Workman and David Kerr, CRC press 1994, Lond on.
  • Ras Oncogene Another group of signal transduction pathway inhibitors are inhibitors of Ras Oncogene.
  • Such inhibitors include inhibitors of farnesyltransferase, geranyl-geranyl transferase, and CAAX proteases as well as anti-sense oligonucleotides, ribozymes and immunotherapy.
  • Such inhibitors have been shown to block ras activation in cells containing wild type mutant ras, thereby acting as anti proliferation agents.
  • Ras oncogene inhibition is discussed in Scharovsky, O.G., Rozados, V.R., Gervasoni, S.I. Matar, P. (2000), Journal of Biomedical Science. 7(4) 292-8 ; Ashby, M.N. (1998), Current Opinion in Lipidology. 9 (2) 99- 102 ; and BioChim. Biophys. Acta, (19899) 1423(3):19-30 .
  • antibody antagonists to receptor kinase ligand binding may also serve as signal transduction inhibitors.
  • This group of signal transduction pathway inhibitors includes the use of humanized antibodies to the extracellular ligand binding domain of receptor tyrosine kinases.
  • Imclone C225 EGFR specific antibody see Green, M.C. et al, Monoclonal Antibody Therapy for Solid Tumors, Cancer Treat.
  • Herceptin ® erbB2 antibody see Tyrosine Kinase Signalling in Breast cancer:erbB Family Receptor Tyrosine Kniases, Breast cancer Res., 2000, 2(3), 176-183
  • 2CB VEGFR2 specific antibody see Brekken, R.A. et al, Selective Inhibition of VEGFR2 Activity by a monoclonal Anti-VEGF antibody blocks tumor growth in mice, Cancer Res. (2000) 60, 5117-5124 ).
  • Non-receptor kinase angiogenesis inhibitors may also be useful in the present invention.
  • Inhibitors of angiogenesis related VEGFR and TIE2 are discussed above in regard to signal transduction inhibitors (both receptors are receptor tyrosine kinases).
  • Angiogenesis in general is linked to erbB2/EGFR signaling since inhibitors of erbB2 and EGFR have been shown to inhibit angiogenesis, primarily VEGF expression. Accordingly, non-receptor tyrosine kinase inhibitors may be used in combination with the compounds of the present invention.
  • anti-VEGF antibodies which do not recognize VEGFR (the receptor tyrosine kinase), but bind to the ligand; small molecule inhibitors of integrin (alpha, betas) that will inhibit angiogenesis; endostatin and angiostatin (non-RTK) may also prove useful in combination with the disclosed compounds.
  • VEGFR the receptor tyrosine kinase
  • small molecule inhibitors of integrin alpha, betas
  • endostatin and angiostatin non-RTK
  • Agents used in immunotherapeutic regimens may also be useful in combination with the compounds of Formula (I).
  • immunologic strategies to generate an immune response. These strategies are generally in the realm of tumor vaccinations.
  • the efficacy of immunologic approaches may be greatly enhanced through combined inhibition of signaling pathways using a small molecule inhibitor. Discussion of the immunologic/tumor vaccine approach against erbB2/EGFR are found in Reilly RT et al. (2000), Cancer Res. 60: 3569-3576 ; and Chen Y, Hu D, Eling DJ, Robbins J, and Kipps TJ. (1998), Cancer Res. 58: 1965-1971 .
  • Agents used in proapoptotic regimens may also be used in the combination of the present invention.
  • Members of the Bcl-2 family of proteins block apoptosis. Upregulation of bcl-2 has therefore been linked to chemoresistance.
  • EGF epidermal growth factor
  • Cell cycle signalling inhibitors inhibit molecules involved in the control of the cell cycle.
  • a family of protein kinases called cyclin dependent kinases (CDKs) and their interaction with a family of proteins termed cyclins controls progression through the eukaryotic cell cycle. The coordinate activation and inactivation of different cyclin/CDK complexes is necessary for normal progression through the cell cycle.
  • CDKs cyclin dependent kinases
  • Several inhibitors of cell cycle signalling are under development. For instance, examples of cyclin dependent kinases, including CDK2, CDK4, and CDK6 and inhibitors for the same are described in, for instance, Rosania et al, Exp. Opin. Ther. Patents (2000) 10(2):215-230 .
  • p21WAF1/CIP1 has been described as a potent and universal inhibitor of cyclindependent kinases (Cdks) ( Ball et al., Progress in Cell Cycle Res., 3: 125 (1997 )).
  • Cdks cyclindependent kinases
  • Compounds that are known to induce expression of p21WAF1/CIP1 have been implicated in the suppression of cell proliferation and as having tumor suppressing activity ( Richon et al., Proc. NatAcad. Sci. U.S.A. 97(18): 10014-10019 (2000 )), and are included as cell cycle signaling inhibitors.
  • Histone deacetylase (HDAC) inhibitors are implicated in the transcriptional activation of p21WAF1/CIP1 ( Vigushin et al., Anticancer Drugs, 13(1): 1-13 (Jan 2002 )), and are suitable cell cycle signaling inhibitors for use herein. Examples of such HDAC inhibitors include:
  • HDAC inhibitors are included in Bertrand European Journal of Medicinal Chemistry 45, (2010) 2095-2116 , particularly the compounds of table 3 therein as indicated below.
  • proteasome inhibitors are drugs that block the action of proteasomes, cellular complexes that break down proteins, like the p53 protein.
  • proteasome inhibitors are marketed or are being studied in the treatment of cancer. Suitable proteasome inhibitors for use herein include:
  • Inhibitors of cancer metabolism Many tumor cells show a markedly different metabolism from that of normal tissues. For example, the rate of glycolysis, the metabolic process that converts glucose to pyruvate, is increased, and the pyruvate generated is reduced to lactate, rather than being further oxidized in the mitochondria via the tricarboxylic acid (TCA) cycle. This effect is often seen even under aerobic conditions and is known as the Warburg Effect.
  • TCA tricarboxylic acid
  • Lactate dehydrogenase A (LDH-A), an isoform of lactate dehydrogenase expressed in muscle cells, plays a pivotal role in tumor cell metabolism by performing the reduction of pyruvate to lactate, which can then be exported out of the cell.
  • the enzyme has been shown to be upregulated in many tumor types.
  • the alteration of glucose metabolism described in the Warburg effect is critical for growth and proliferation of cancer cells and knocking down LDH-A using RNA-i has been shown to lead to a reduction in cell proliferation and tumor growth in xenograft models.
  • Inhibitors of cancer metabolism including inhibitors of LDH-A, are suitable for use in combination with the compounds of this invention.
  • dry blend and derivatives thereof, as used herein refers to formulated particles that comprise Compound A and/or diluents and/or binders and/or lubricants and/or disintegrants such that the particles are suitable for utilization in preparing solid oral pharmaceutical dosage forms, suitably tablets, suitably capsules and are produced by dry blending or dry granulation. It is possible to administer the dry blend directly to a subject in need thereof as a medicament. However, it is anticipated that the dry blend are most appropriately utilized in the preparation of solid oral pharmaceutical dosage forms, suitably tablets, suitably capsules, as indicated above.
  • solid oral pharmaceutical dosage form and “solid dosage form” and derivatives thereof, as used herein, unless otherwise defined, refers to a final pharmaceutical preparation that comprises Compound A, such as: tablets, capsules, pellets, lozenges, sachets and powders (including coated versions of any of such preparations), suitably tablets, suitably capsules, that are suitable for in vivo administration.
  • Compound A such as: tablets, capsules, pellets, lozenges, sachets and powders (including coated versions of any of such preparations), suitably tablets, suitably capsules, that are suitable for in vivo administration.
  • an excipient for use herein is substantially free of water
  • the excipient could contain minor amounts of water, for example: about 5% by weight or less, suitably about 2.5% by weight of less, suitably about 1% by weight of less.
  • very minor amounts of water can be in the excipient component without adversely affecting the performance of the solid dosage from, suitably the tablet, suitably the capsule.
  • the tablets of the present invention comprise Compound A, a diluent (also known as filler or bulking agent), and suitably also a binder and/or a lubricant and/or a disintegrant.
  • a diluent also known as filler or bulking agent
  • a binder and/or a lubricant and/or a disintegrant may provide one or more functions in the tablet formulation, although the material is usually included for a primary function.
  • the percentages of diluent, binder, lubricant and disintegrant provided herein and in the claims are by weight of the tablet.
  • Diluents provide bulk, for example, in order to make the tablet a practical size for processing. Diluents may also aid processing, for example, by providing improved physical properties such as flow, compressibility, and tablet hardness. Because of the relatively high percentage of diluent and the amount of direct contact between the diluent and the active compound in the typical pharmaceutical formulation, the interaction of the diluent with the active compound is of particular concern to the formulator.
  • diluents suitable for use in the present invention include either the following or an anhydrous version thereof: water-soluble fillers and water-insoluble fillers, such as calcium phosphate (e.g., di and tri basic, hydrated or anhydrous), calcium sulfate, calcium carbonate, magnesium carbonate, kaolin, lactose that is substantially free of water, suitably spray dried or anhydrous lactose (collectively lactose as used herein), cellulose (e.g., microcrystalline cellulose, powdered cellulose), pregelatinized starch, starch, lactitol, mannitol, sorbitol, maltodextrin, powdered sugar, compressible sugar, sucrose, dextrose, and inositol. Diluents that are substantially free of water are suitable for tablets of the current invention. In one embodiment of the present invention, the diluent is composed of one or both of Mannitol and microcrystalline cellulose.
  • the tablets of the present invention typically comprise from about 25% to about 89%, of one or more diluents.
  • One aspect of the present invention comprises pharmaceutical tablets, wherein the tablets are formulated using a diluent or diluents that are substantially free of water.
  • Binders impart cohesive properties to the powdered material.
  • binders suitable for use in the present invention include either the following or an anhydrous version thereof: hydroxypropyl methyl cellulose (HPMC), hydroxypropyl cellulose (HPC), hydroxyethyl cellulose (HEC) and ethyl cellulose (EC)], polyvinylpyrrolidone. Binders that are substantially free of water are suitable for tablets of the current invention. In one embodiment of the present invention, the binder is hydroxypropyl methyl cellulose (HPMC) or Hypromellose.
  • the tablets of the present invention typically comprise up to about 2 - 8% binder such as about 2%, about 3%, about 4% , about 5%, about 6% about 7%, and about 8% w/w.
  • the tablets suitably comprise up to about 5% binder.
  • One aspect of the present invention comprises pharmaceutical tablets, wherein the tablets are formulated using a binder or binders that are substantially free of water.
  • Lubricants are generally used to enhance processing, for example, to prevent adhesion of the formulation material to manufacturing equipment, reduce interparticle friction, improve rate of flow of the formulation, and/or assist ejection of the formulations from the manufacturing equipment.
  • lubricants suitable for use in the present invention include either the following or an anhydrous version thereof: talc, stearates (e.g., magnesium stearate, calcium stearate, zinc stearate, palmitostearate), stearic acid, hydrogenated vegetable oils, glyceryl behanate, polyethylene glycol, ethylene oxide polymers (e.g., CARBOWAXes), liquid paraffin, sodium lauryl sulfate, magnesium lauryl sulfate, sodium oleate, sodium stearyl fumarate, DL-leucine, and silica derivatives (e.g., colloidal silicon dioxide, colloidal silica, pyrogenic silica, and sodium silicoaluminate).
  • the tablets of the present invention typically comprise up to about 2% lubricant.
  • the tablets suitably comprise up to about 1 %, suitably up to about 0.75% lubricant.
  • One aspect of the present invention comprises pharmaceutical tablets, wherein the tablets are formulated using a lubricant or lubricants that are substantially free of water.
  • Disintegrants are employed to facilitate breakup or disintegration of the formulation after administration.
  • examples of disintegrants suitable for use in the present invention include either the following or an anhydrous version thereof: starches, celluloses, gums, crosslinked polymers, and effervescent agents, such as corn starch, potato starch, pregelatinized starch, modified corn starch, croscarmellose sodium, crospovidone, sodium starch glycolate, Veegum HV, methyl cellulose, microcrystalline cellulose, cellulose, colloidal silicon dioxide, modified cellulose gum (e.g., Ac-Di-Sol R), agar, bentonite, montmorillonite clay, natural sponge, cation exchange resins, ion exchange resins (e.g., polyacrin potassium), alginic acid and alginates, guar gum, citrus pulp, carboxymethylcellulose and salts thereof such as sodium lauryl sulfate, magnesium aluminum silicate, hydrous aluminum silicate, sodium bicarbonate in admi
  • the tablets of the present invention typically comprise an amount from 2 % to about 5% disintegrant, suitably about 2%, about 3% , about 4%, and about 5% w/w.
  • the tablets suitably comprise about 3% disintegrant.
  • One aspect of the present invention comprises pharmaceutical tablets, wherein the tablets are formulated using a disintegrant or disintegrants that are substantially free of water.
  • each pharmaceutical compound When administered in vivo, each pharmaceutical compound acts uniquely in regards to therapeutic drug levels. Further, pharmaceutically active compounds are often associated with undesirable side effects such as; toxicity (e.g. genotoxicity, teratogenicity) and undesirable physical or psychological manifestations.
  • toxicity e.g. genotoxicity, teratogenicity
  • undesirable physical or psychological manifestations e.g. genotoxicity, teratogenicity
  • the drug In addition to balancing the drug's chemical properties with those of the excipients, the drug must be administered at a specific amount that is sufficient to provide the desired therapeutic drug level but less than the amount that presents an unacceptable side effect profile, or within a therapeutic window for that particular drug.
  • One embodiment of this invention is directed to administering Compound A in an amount sufficient to provide the desired therapeutic effect and an acceptable side effect profile.
  • Preliminary PK data was obtained in subjects with solid tumors following single- and repeat-dose oral administration of Compound tablets.
  • Plasma concentrations for Compound were not measurable for all subjects over the 24-hour time period, especially for subjects administered low doses ranging from 0.125-0.50 mg.
  • AUC (0-24) and Cmax values were dose proportional up to 6 mg, lower than dose proportional following 8 mg and greater than dose proportional following the 10 mg dose.
  • Most subjects had samples taken up to 24 hrs following single dose and half-life and AUC(0-inf) could not be calculated.
  • Median Tmax ranged from 1.0 to 3.0 hours.
  • Mean trough concentrations after repeat dosing up to 10 cycles ranged from 10.0 to 18.9 ng/mL following 2.0 mg daily and from 7.8 to 17.3 ng/mL following 2.5 mg.
  • Mean (CV%) trough concentrations on Day 15 after repeat dosing of 2.5mg of Compound were 16.8ng/mL (54%) and ranged from 0.68 to 49.0 ng/mL.
  • a dose of 2 mg administered once daily was selected based on tolerability, exposure-response relationship with pharmacodynamic markers in tumor biopsies, and clinical activity.
  • a 0.5 mg dose was also selected to accommodate lower strength dosing, for example when used in combination with another anti-neoplastic compound or when dose reduction is required due to toxicity.
  • composition of coatings studied is given in Table 7.
  • Opadry ® white provided the least amount of protection for 1mg strength, as the observed impurity levels appear to be similar to the uncoated tablets.
  • Two different formulations of Opadry ® pink and Opadry ® yellow were tested and all provided adequate light protection.
  • Table 9 Structures of Impurities Code Name Structural Formula Impurity I (RRT 0.81) Impurity II (RRT 0.87) RRT 0.89 Structure is not determined
  • the color in the tablet coating appears to play an important role in protecting Compound A from light degradation.
  • the color coatings may prevent light induced impurities from being formed either by physically blocking the damaging wavelength or by providing chemical protection (scavenging oxidizing radicals).
  • uncoated tablets, wax coated tablets, white coated tablets and the like can be placed in a protective light resistant blister pack or light resistant bottle to keep the drug away from light.
  • micronized drug substance is used with a specified disintegrant or specified disintegrants.
  • a suspension formulation was prepared from micronized drug substance suspended in a vehicle consisting of 5 % mannitol, 1.5 % hypromellose and 0.2 % sodium lauryl sulfate ("micronized” in Table 10 below). Two concentrations were prepared: 26.9 mcg/mL and 134.4 mcg/mL and dosed the same day. On the third day the 134.4 mcg/mL suspension was dosed again along with a freshly prepared suspension of similar concentration Table 10 summarizes the exposure data (AUC) and Figure 1 represents the results of the pre-clinical studies.
  • the micronized drug substance is in a formulation with a specified disintegrant or specified disintegrants, suitably selected from one or more of: sodium lauryl sulfate, colloidal silicon dioxide and croscamellose sodium.
  • micronized is meant the standard usage in the art that the drug particles are processed, for example by milling, bashing and/or grinding, to significantly reduce particle size over those produced naturally during chemical synthesis.
  • at least 50% of the subject particles are 30 micron or less, suitably at least 50% of the particles are 10 micron or less, suitably at least 50% of the particles are 5 micron or less.
  • a suitable particle size distribution for the drug particles of the invention are as follows.
  • wet granulation or a tabletting technique that uses a significant water concentration is unsuitable for preparing tablets of Compound A, particularly on a commercial scale, because upon contact with water during the formulation process, Compound A can revert to Compound B which is significantly less soluble.
  • Experiments were undertaken to determine the acceptable level of the desolvated Compound B in a pharmaceutical dosage form and the appropriate formulation techniques.
  • Table 12 Stability data for 1 mg tablets containing Compound A, made generally according to Example 2, exposed to four different storage conditions (30's count in HDPE Bottles with Desiccant) is presented in Table 12 and Figure 2 .
  • Table 12 Storage Condition Time (Months) Test Content (% Label Claim) DMSO Content (% of Compound A % Desolvation (Calculated) * Dissolution (% released) Mean (min- max) Specification 90.0-110.0% 9.0 - 13.5 NMT 20% Complies with USP Q 75% at 30 min.
  • the DMSO content lower limit corresponds to about 20% desolvation.
  • Initial DMSO content corresponded to about 0%.
  • the initial DMSO content will be less than about 2% desolvation, suitably less than about 4% desolvation, suitably less than about 8% desolvation.
  • the DMSO content will not be less than an equivalent of about 5% desolvation during the shelf life of the tablet, suitably about 10% desolvation suitably about 15% desolvation, suitably about 20% desolvation. Consequently, dry direct compression and dry granulation were found to be appropriate formulation techniques.
  • the tablets of the present invention will typically be sized up to 1 gram, suitably from about 140mg to about 175mg. These tablets will contain Compound A in an amount selected from: 0.5mg, 1mg and 2mg, by weight of Compound B. Tablet formulations of the invention may have a variety of shapes, including: round, modified round, diamond, modified capsule, modified oval, oval and hexagonal, and may optionally have a tilt.
  • excipients and tabletting technique employed depends on the further properties of Compound A and the excipients, e.g., compressibility, flowability, particle size, compatibility, and density.
  • the tablets may be prepared according to methods known in the art, including dry direct compression and dry granulation, and the type of excipients used will vary depending on the exact process employed. It has been found that dry direct compression is particularly suitable for providing high strength, low breakage tablets comprising relatively low concentrations of Compound A (e.g., less than about 1.5 %, suitably less than 1 %), on a scale suitable for commercial production.
  • Suitable dry direct compression tablets of the invention comprise dry blend comprising Compound A and one or more of fillers, binders and disintegrants, mixed with additional filler, binder, disintegrant and/or lubricant to form a compression mixture that is compressed to form tablets.
  • compositions in tablet form suitably prepared on a commercial scale, that comprise Compound A, wherein the tablet is made by a dry direct compression process using a diluent or diluents that are substantially free water.
  • Such tablets that contain a film coat, wherein the film coat contains a colored pigment.
  • tablets that comprise Compound A, wherein the tablet is made by a dry direct compression process, suitably on a commercial scale, using a diluent or diluents that are substantially free water and the Compound A particles are micronized.
  • the micronization of compound A enhances the biological exposure by increasing the particle specific surface area, as well as providing adequate content uniformity of the low strength solid dosage form.
  • a surfactant as disclosed herein further enhances the biological exposure by increasing the wettability of the micronized compound A.
  • the tablets of the present invention comprise:
  • the diluent is suitably a combination of mannitol and microcrystalline cellulose
  • the binder is suitably HPMC
  • the lubricant is suitably magnesium stearate
  • the disintegrant is suitably a combination of sodium lauryl sulfate, colloidal silicon dioxide and croscamellose sodium.
  • tablets are coated with a film coat formed from an aqueous film coat composition.
  • Aqueous film coat compositions suitable for use in the present invention comprise a film-forming polymer, water as a vehicle, and optionally one or more adjuvants such as are known in the film-coating art.
  • the film coat will contain a colored pigment.
  • the colored pigment contains iron oxide.
  • the film-forming polymer is selected to form coatings with mechanical properties (e.g., mechanical strength, flexibility) suitable to meet performance requirements, such as those required by the intended use environment (e.g., dissolution profile in gastrointestinal fluids), and/or use (e.g. solution viscosity).
  • mechanical properties e.g., mechanical strength, flexibility
  • performance requirements such as those required by the intended use environment (e.g., dissolution profile in gastrointestinal fluids), and/or use (e.g. solution viscosity).
  • suitable film-forming polymers include cellulosic polymers (e.g., cellulose ethers such as HPMC, HPC, MC, EC, HEC, CAP, sodium ethyl cellulose sulfate, carboxymethyl cellulose and the like); polyvinylpyrolidone; zein; and acrylic polymers (e.g., methacrylic acid/methacrylic acid ester copolymers such as methacrylic acid/methylmethacrylate copolymers and the like).
  • Cellulosic polymers are preferred in the present invention, especially cellulosic ethers and more especially HPMC and HPC.
  • the polymers are typically provided in either aqueous or organic solvent based solutions or aqueous dispersions.
  • the polymers may be provided in dry form, alone or in a powdery mixture with other components (e.g., a plasticizer and/or colorant), which is made into a solution or dispersion by the user by admixing with the aqueous vehicle.
  • the aqueous film coat composition further comprises water as a vehicle for the other components, to facilitate their delivery to the tablet surface.
  • vehicle may optionally further comprise one or more water soluble solvents, e.g., alcohols (e.g., methanol, isopropanol, propanol) and ketones (e.g., acetone).
  • alcohols e.g., methanol, isopropanol, propanol
  • ketones e.g., acetone
  • the aqueous film coating composition may optionally comprise one or more adjuvants known in the art, such as plasticizers, colorants, detackifiers, secondary film-forming polymers, flow aids, surfactants (e.g., to assist spreading), maltodextrin, and polydextrose.
  • adjuvants known in the art, such as plasticizers, colorants, detackifiers, secondary film-forming polymers, flow aids, surfactants (e.g., to assist spreading), maltodextrin, and polydextrose.
  • Plasticizers provide flexibility to the film, which may reduce film cracking and improve adhesion to the tablet. Suitable plasticizers will generally have a high degree of compatibility with the film-forming polymer and sufficient permanence such that the coating properties are generally stable.
  • suitable plasticizers include glycerin, propylene glycol, polyethylene glycols (e.g., molecular weight from 200 to 20,000, including Union Carbide's PEG 400, 4000, 6000, 8000, and 20,000), glycerin triacetate (aka triacetin), acetylated monoglyceride, citrate esters (e.g., triethyl citrate, acetyl triethyl citrate, tributyl citrate, acetyl tributyl citrate), phthalate esters (e.g., diethyl phthalate), mineral oil and hydrogenated glucose syrup.
  • the plasticizer is chosen from polyethylene glycols, triacetin, propy
  • Compound A was found to be sensitive to photo-induced degradation.
  • a film coating suitably a colored film coating, is advantageous to improve stability.
  • the aqueous film coat composition will typically comprise from about 5% to about 25%, suitably about 5% to about 20%, coating solids in the vehicle.
  • the solids typically comprise from about 25% to about 70%, suitably about 60% to about 70% film-forming polymer, about 5% to about 10%, suitably about 6% to about 8%, plasticizer, and about 20% to about 35% pigment and colorant, by weight.
  • the colorant comprises from about 0.5 to 3% by weight.
  • aqueous film coating compositions are commercially available.
  • the aqueous film coat composition may be provided in the form of a solution or dispersion.
  • the composition may be provided in a dry form that can be combined with the vehicle components according to supplier instructions prior to coating the tablet.
  • aqueous film coating compositions are those commercially available from Colorcon, Inc. of West Point, PA, under the trade name OPADRY and OPADRY II (nonlimiting examples include Opadry Pink YS-1-14762-A or 03B140009, Opadry Yellow YS-1-12525-A or 03B120006). These compositions are available as dry film coating compositions that can be diluted in water shortly before use.
  • the tablets are also suitably coated to provide a uniform coating without speckling.
  • the tablets are typically coated to provide a dry tablet weight gain of from about 2 to about 5%, suitably about 2.5 to 4%.
  • the uncoated tablet cores are coated with the aqueous film coating composition by methods well known in the art using commercially available equipment (e.g., Thomas Accela-Cota, Vector Hi-Coater, Compu-Lab 36).
  • the process usually involves rolling or tumbling the tablets in a pan, or suspending the tablets on a cushion of air (fluidized bed), and intermittently or continuously (preferably continuously) spraying a fine mist of atomized droplets of the coating composition onto the tablets, the droplets wetting, spreading and coalescing on the surface of the tablets to form an adherent and coherent film coating.
  • the tablets are typically heated to about 40 to 50°C, suitably about 45 to 50°C, e.g., by air having a temperature of up to about 85°C, suitably about 65 to 80°C.
  • the invented tablets may be administered in therapeutically effective amounts to treat or prevent a disease state, e.g., as described in the above referenced International Application No. PCT/JP2005/011082 , and United States Patent Publication No. US 2006/0014768 .
  • Also disclosed is a method of inhibiting MEK activity in humans comprising administering to a subject in need of such activity a therapeutically effective amount of a solid oral pharmaceutical dosage form of the present invention.
  • Compound A in the manufacture of a solid oral pharmaceutical dosage form of the present invention for use in treating cancer.
  • Compound A in the manufacture of a solid oral pharmaceutical dosage form of the present invention for use in inhibiting MEK.
  • Solid oral pharmaceutical dosage form for use as a MEK inhibitor which comprises Compound A and a pharmaceutically acceptable carrier of the present invention.
  • a solid oral pharmaceutical dosage form for use in the treatment of cancer which comprises Compound A and a pharmaceutically acceptable carrier of the present invention.
  • Solid oral pharmaceutical dosage form for use in inhibition MEK which comprises Compound A and a pharmaceutically acceptable carrier of the present invention.
  • micronized drug substance sodium lauryl sulfate, silicon dioxide, croscarmellose sodium, microcrystalline cellulose and hypromellose are screened, if required, and transferred into a suitable bin blender and blended.
  • the magnesium stearate is screened, if required, transferred to the bin blender and blended for an additional time.
  • the lubricated blend is compressed on a rotary tablet press to the target weight for each strength (145 mg, 155 mg and 165 mg corresponding to 0.5 mg, 1 mg and 2 mg, respectively).
  • the compressed tablets are sampled for in-process monitoring of individual weight variation, appearance, hardness, thickness, friability and disintegration time.
  • Tablet cores are sprayed with an aqueous suspension of Opadry ® Pink YS-1-14762-A) (for 2 mg strength), Opadry ® Yellow YS-1-12525-A (for 0.5 mg strength) or Opadry ® White OY-S-28876 (for 1 mg strength). Coating continues until a target weight gain of approximately 3% is attained. The tablets are then dried and bulk packed into HDPE containers with plastic liners and desiccant bags, and stored until packaged.
EP22209104.3A 2010-12-20 2011-12-20 Novel pharmaceutical composition Active EP4159204B1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201061424967P 2010-12-20 2010-12-20
PCT/US2011/066021 WO2012088033A2 (en) 2010-12-20 2011-12-20 Novel pharmaceutical composition
EP20179788.3A EP3808343A1 (en) 2010-12-20 2011-12-20 Novel pharmaceutical composition
EP11849974.8A EP2654736B1 (en) 2010-12-20 2011-12-20 Novel pharmaceutical composition

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
EP11849974.8A Division EP2654736B1 (en) 2010-12-20 2011-12-20 Novel pharmaceutical composition
EP20179788.3A Division EP3808343A1 (en) 2010-12-20 2011-12-20 Novel pharmaceutical composition

Publications (2)

Publication Number Publication Date
EP4159204A1 EP4159204A1 (en) 2023-04-05
EP4159204B1 true EP4159204B1 (en) 2024-02-14

Family

ID=46314827

Family Applications (4)

Application Number Title Priority Date Filing Date
EP22209107.6A Active EP4159205B1 (en) 2010-12-20 2011-12-20 Novel pharmaceutical composition
EP11849974.8A Active EP2654736B1 (en) 2010-12-20 2011-12-20 Novel pharmaceutical composition
EP22209104.3A Active EP4159204B1 (en) 2010-12-20 2011-12-20 Novel pharmaceutical composition
EP20179788.3A Pending EP3808343A1 (en) 2010-12-20 2011-12-20 Novel pharmaceutical composition

Family Applications Before (2)

Application Number Title Priority Date Filing Date
EP22209107.6A Active EP4159205B1 (en) 2010-12-20 2011-12-20 Novel pharmaceutical composition
EP11849974.8A Active EP2654736B1 (en) 2010-12-20 2011-12-20 Novel pharmaceutical composition

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP20179788.3A Pending EP3808343A1 (en) 2010-12-20 2011-12-20 Novel pharmaceutical composition

Country Status (36)

Country Link
US (5) US8580304B2 (zh)
EP (4) EP4159205B1 (zh)
JP (2) JP6126014B2 (zh)
KR (1) KR101911109B1 (zh)
CN (1) CN103998041B (zh)
AR (2) AR084102A1 (zh)
AU (1) AU2011349422B2 (zh)
BR (1) BR112013015602B1 (zh)
CA (1) CA2822701C (zh)
CL (1) CL2013001779A1 (zh)
CR (1) CR20130352A (zh)
CY (1) CY1123376T1 (zh)
DK (1) DK2654736T3 (zh)
DO (1) DOP2013000138A (zh)
EA (1) EA025198B1 (zh)
ES (1) ES2820536T3 (zh)
FI (2) FI4159205T3 (zh)
HR (1) HRP20201409T1 (zh)
HU (1) HUE050788T2 (zh)
IL (1) IL226855A (zh)
JO (1) JO3594B1 (zh)
LT (1) LT2654736T (zh)
MA (1) MA34883B1 (zh)
MX (1) MX2013007073A (zh)
MY (1) MY170501A (zh)
NZ (1) NZ612157A (zh)
PE (1) PE20140040A1 (zh)
PL (1) PL2654736T3 (zh)
PT (1) PT2654736T (zh)
SG (1) SG191054A1 (zh)
SI (1) SI2654736T1 (zh)
TW (1) TWI505828B (zh)
UA (1) UA113158C2 (zh)
UY (1) UY33818A (zh)
WO (1) WO2012088033A2 (zh)
ZA (1) ZA201304189B (zh)

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20150038068A (ko) 2012-08-17 2015-04-08 에프. 호프만-라 로슈 아게 코비메티닙 및 베무라피닙을 투여함을 포함하는 흑색종의 조합 치료법
CA2891346A1 (en) * 2012-11-30 2014-06-05 Gossett Campbell Pharmaceutical composition comprising trametinib dimethyl sulfoxide
WO2015019125A1 (en) * 2013-08-05 2015-02-12 Societe De Developpement Et De Recherche Industrielle Molecular targets for the treatment of wounds, in particular chronic wounds
CN103819471A (zh) * 2013-11-25 2014-05-28 镇江圣安医药有限公司 吡啶并(4,3-d)嘧啶-1(2H)-基苯基乙酰胺的衍生物及其应用
WO2015081566A1 (zh) * 2013-12-06 2015-06-11 杭州普晒医药科技有限公司 曲美替尼及其溶剂化物的晶型、其制备方法、含有它们的药物组合物及其用途
EP2913048A1 (en) 2014-02-27 2015-09-02 ratiopharm GmbH Pharmaceutical composition comprising trametinib
KR101545268B1 (ko) 2015-02-05 2015-08-20 보령제약 주식회사 정제 및 이의 제조방법
WO2016126012A1 (en) * 2015-02-05 2016-08-11 Boryung Pharmaceutical Co., Ltd Tablet and method of preparing the same
US20170020880A1 (en) * 2015-07-22 2017-01-26 Hetero Research Foundation Pharmaceutical compositions of trametinib
TW202329972A (zh) * 2016-08-10 2023-08-01 瑞士商赫孚孟拉羅股份公司 包含Akt蛋白質激酶抑制劑之醫藥組合物
RU2627692C1 (ru) 2016-10-10 2017-08-10 Закрытое акционерное общество "Р-Фарм" (ЗАО "Р-Фарм") N-{ 3-[3-циклопропил-5-(2-фторо-4-иодофениламино)-6,8-диметил-2,4,7-триоксо-3,4,6,7-тетрагидро-2Н-пиридо[4,3-d]пиримидин-1-ил]-фенил} -циклопропанкарбоксамида диметилсульфоксида сольват в качестве ингибитора МЕК1/2
JP2019535696A (ja) 2016-11-25 2019-12-12 ジエンス ヘンルイ メデイシンカンパニー リミテッドJiangsu Hengrui Medicine Co.,Ltd. ピリドン誘導体の医薬組成物およびその製造方法
PT3618875T (pt) 2017-05-02 2023-08-07 Novartis Ag Terapia de combinação que compreende um inibidor de raf e trametinib
RU2749719C1 (ru) * 2020-10-08 2021-06-16 Нестерук Владимир Викторович Препарат для лечения анемии, связанной с хроническим заболеванием почек, и способ его получения
WO2023105286A1 (en) 2021-12-06 2023-06-15 My Personal Therapeutics Ltd A combination treatment for cancer

Family Cites Families (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS61205257A (ja) * 1985-03-08 1986-09-11 Ishihara Sangyo Kaisha Ltd ベンゾイルウレア系化合物、その製法及びそれを含有する抗ガン剤
US4727077A (en) 1985-02-20 1988-02-23 Ishihara Sangyo Kaisha Ltd. Benzoyl urea compounds, process for their production, and antitumorous compositions containing them
US5559235A (en) 1991-10-29 1996-09-24 Glaxo Wellcome Inc. Water soluble camptothecin derivatives
US5342947A (en) 1992-10-09 1994-08-30 Glaxo Inc. Preparation of water soluble camptothecin derivatives
US5681835A (en) 1994-04-25 1997-10-28 Glaxo Wellcome Inc. Non-steroidal ligands for the estrogen receptor
US5491237A (en) 1994-05-03 1996-02-13 Glaxo Wellcome Inc. Intermediates in pharmaceutical camptothecin preparation
GB9716557D0 (en) 1997-08-06 1997-10-08 Glaxo Group Ltd Benzylidene-1,3-dihydro-indol-2-one derivatives having anti-cancer activity
US6825180B2 (en) 2001-05-18 2004-11-30 Cell Therapeutics, Inc. Pyridopyrimidine compounds and their uses
BRPI0511967B8 (pt) * 2004-06-11 2021-05-25 Japan Tobacco Inc derivados de 5-amino-2,4,7-trioxo-3,4,7,8-tetrahidro-2h-pirido[2,3-d] pirimidina, seu uso e composição farmacêutica que os compreende
US7378423B2 (en) 2004-06-11 2008-05-27 Japan Tobacco Inc. Pyrimidine compound and medical use thereof
US8648116B2 (en) * 2005-07-21 2014-02-11 Ardea Biosciences, Inc. Derivatives of N-(arylamino) sulfonamides including polymorphs as inhibitors of MEK as well as compositions, methods of use and methods for preparing the same
WO2008048469A2 (en) * 2006-10-13 2008-04-24 Actavis Group Pct Hf Controlled-release coated dosage forms containing galantamine
WO2008060890A2 (en) * 2006-11-09 2008-05-22 Gilead Colorado, Inc. Darusentan oral dosage form
JO2985B1 (ar) * 2006-12-20 2016-09-05 Takeda Pharmaceuticals Co مثبطات كينازmapk/erk
UY30892A1 (es) 2007-02-07 2008-09-02 Smithkline Beckman Corp Inhibidores de la actividad akt
ECSP077628A (es) * 2007-05-03 2008-12-30 Smithkline Beechman Corp Nueva composición farmacéutica
US20090087489A1 (en) * 2007-09-25 2009-04-02 Bella Gerber Imatinib compositions
CN104530061B (zh) * 2008-07-08 2017-05-10 贝达药业股份有限公司 埃克替尼盐酸盐晶型、药物组合物和用途
EA201270453A1 (ru) 2009-09-23 2013-12-30 ГЛЭКСОСМИТКЛАЙН ЭлЭлСи Комбинация
KR20120099219A (ko) 2009-09-23 2012-09-07 글락소스미스클라인 엘엘씨 조합물
EP2482819A4 (en) * 2009-09-28 2013-02-20 Glaxosmithkline Llc COMBINATION
PL2488033T3 (pl) * 2009-10-16 2019-12-31 Novartis Ag Kombinacja zawierająca inhibitor MEK i inhibitor B-raf
CA2781063A1 (en) 2009-11-17 2011-05-26 Glaxosmithkline Llc Combination
US20130165456A1 (en) 2010-08-26 2013-06-27 Tona M. Gilmer Combination
US8977217B1 (en) 2013-02-20 2015-03-10 Triquint Semiconductor, Inc. Switching device with negative bias circuit

Also Published As

Publication number Publication date
MA34883B1 (fr) 2014-02-01
AR122185A2 (es) 2022-08-24
EA025198B1 (ru) 2016-11-30
UA113158C2 (xx) 2016-12-26
ZA201304189B (en) 2014-02-26
WO2012088033A3 (en) 2014-03-13
EA201390913A1 (ru) 2014-04-30
EP2654736A2 (en) 2013-10-30
CN103998041B (zh) 2016-08-17
CN103998041A (zh) 2014-08-20
US9399021B2 (en) 2016-07-26
PT2654736T (pt) 2020-09-24
US20120183613A1 (en) 2012-07-19
TW201249441A (en) 2012-12-16
EP3808343A1 (en) 2021-04-21
AU2011349422B2 (en) 2015-12-10
HUE050788T2 (hu) 2021-01-28
KR20130130028A (ko) 2013-11-29
NZ612157A (en) 2015-05-29
CY1123376T1 (el) 2021-12-31
HRP20201409T1 (hr) 2020-11-27
US8580304B2 (en) 2013-11-12
EP4159204A1 (en) 2023-04-05
AU2011349422A1 (en) 2013-05-02
JP6126014B2 (ja) 2017-05-10
AR084102A1 (es) 2013-04-24
TWI505828B (zh) 2015-11-01
SG191054A1 (en) 2013-08-30
KR101911109B1 (ko) 2018-10-23
US20140099365A1 (en) 2014-04-10
CA2822701A1 (en) 2012-06-28
BR112013015602A2 (pt) 2017-02-21
WO2012088033A2 (en) 2012-06-28
ES2820536T3 (es) 2021-04-21
MY170501A (en) 2019-08-08
JP2017137299A (ja) 2017-08-10
PL2654736T3 (pl) 2020-12-14
MX2013007073A (es) 2013-09-26
US20140037726A1 (en) 2014-02-06
BR112013015602B1 (pt) 2022-03-03
EP4159205A1 (en) 2023-04-05
JP2014510704A (ja) 2014-05-01
FI4159205T3 (fi) 2024-04-23
EP2654736B1 (en) 2020-07-01
CA2822701C (en) 2018-10-23
US20130266649A1 (en) 2013-10-10
LT2654736T (lt) 2020-09-25
US9271941B2 (en) 2016-03-01
PE20140040A1 (es) 2014-02-26
CL2013001779A1 (es) 2014-06-27
DK2654736T3 (da) 2020-09-14
IL226855A (en) 2017-04-30
CR20130352A (es) 2013-12-18
UY33818A (es) 2012-07-31
EP2654736A4 (en) 2015-04-22
EP4159205B1 (en) 2024-02-14
US20140154316A1 (en) 2014-06-05
FI4159204T3 (fi) 2024-04-22
US9155706B2 (en) 2015-10-13
JO3594B1 (ar) 2020-07-05
DOP2013000138A (es) 2013-11-30
SI2654736T1 (sl) 2020-10-30

Similar Documents

Publication Publication Date Title
EP4159204B1 (en) Novel pharmaceutical composition
US20170189408A1 (en) Novel pharmaceutical composition
AU2023266278A1 (en) Novel pharmaceutical composition

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN PUBLISHED

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

AC Divisional application: reference to earlier application

Ref document number: 2654736

Country of ref document: EP

Kind code of ref document: P

Ref document number: 3808343

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

17P Request for examination filed

Effective date: 20230324

RBV Designated contracting states (corrected)

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230514

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

RAX Requested extension states of the european patent have changed

Extension state: ME

Payment date: 20230727

Extension state: BA

Payment date: 20230727

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40086699

Country of ref document: HK

17Q First examination report despatched

Effective date: 20230818

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

INTG Intention to grant announced

Effective date: 20231219

AC Divisional application: reference to earlier application

Ref document number: 2654736

Country of ref document: EP

Kind code of ref document: P

Ref document number: 3808343

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

RIN2 Information on inventor provided after grant (corrected)

Inventor name: WANG, LIHONG

Inventor name: HENRIQUEZ, FRANCISCO

Inventor name: LE, NGOCDIEP T.

Inventor name: DEMARINI, DOUGLAS, J.

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602011074588

Country of ref document: DE

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D